Abstract Background Perioperative chemotherapy is the standard of care for patients with locally advanced gastric and gastroesophageal junction cancer. Recent evidence demonstrated the addition of programmed cell death protein 1 (PD-1) inhibitors enhanced therapeutic efficacy. However, the mechanisms of response and resistance remain largely undefined. A detailed multiomic investigation is essential to elucidate these mechanisms. Methods We performed whole-exome sequencing, whole-transcriptome sequencing, multiplex immunofluorescence and single-cell RNA sequencing on matched pretreatment and post-treatment samples from 30 patients enrolled in an investigator-initiated Phase 2 clinical trial ([55]NCT04908566). All patients received neoadjuvant PD-1 inhibitors in combination with chemotherapy. A major pathologic response (MPR) was defined as the presence of no more than 10% residual viable tumor cells following treatment. Results Before treatment, the positive ratio of CD3+T cells in both the tumor parenchyma and stroma was significantly higher in the non-MPR group compared with the MPR group (p=0.042 and p=0.013, respectively). Least absolute shrinkage and selection operator regression was employed for feature gene selection and 13 genes were ultimately used to construct a predictive model for identifying MPR after surgery. The model exhibited a perfect area under curve (AUC) of 1.000 (95% CI: 1.000 to 1.000, p<0.001). Post-treatment analysis revealed a significant increase in CD3+T cells, CD8+T cells and NK cells in the tumor stroma of MPR patients. In the tumor parenchyma, aside from a marked increase in CD8+T cells and NK cells, a notable reduction in macrophage was also observed (all p<0.05). Importantly, forkheadbox protein 3 (FOXP3), the principal marker for regulatory T cells (Treg) cells, showed a significant decrease during treatment in MPR patients. FOXP3 expression in the non-MPR group was significantly higher than in the MPR group (p=0.0056) after treatment. Furthermore, single-cell RNA sequencing analysis confirmed that nearly all Treg cells were derived from the non-MPR group. Conclusions Our study highlights the critical role of dynamic changes within the tumor immune microenvironment in predicting the efficacy of neoadjuvant combined immunochemotherapy. We examined the disparities between MPR/non-MPR groups, shedding light on potential mechanisms of immune response and suppression. In addition to bolstering cytotoxic immune responses, specifically targeting Treg cells may be crucial for enhancing treatment outcomes. Keywords: Gastric Cancer, Neoadjuvant, Immunotherapy, Tumor microenvironment - TME __________________________________________________________________ Key messages. * The addition of immunotherapy to perioperative chemotherapy enhances therapeutic efficacy, but the mechanisms of response and resistance remain unclear. We performed multiomic investigation on matched pretreatment and post-treatment tissue samples from 30 patients with locally advanced gastric and gastroesophageal junction cancer. Our study highlights the critical role of dynamic changes within the tumor immune microenvironment in predicting the efficacy of immunotherapy combined with chemotherapy. In addition to bolstering cytotoxic immune responses, specifically targeting regulatory T cell cells may be crucial for enhancing treatment outcomes. Introduction Gastric and gastroesophageal junction (G/GEJ) cancer is the fifth most common cancer and the fourth most lethal cancer worldwide.[56]^1 G/GEJ adenocarcinoma accounts for 95% of G/GEJ cancers. For patients with locally advanced G/GEJ adenocarcinoma, perioperative chemotherapy combined with surgical resection is recommended. This combined treatment approach maximizes systemic therapy while reducing the stage of locally advanced tumors and improving the eradication of residual disease and prevention of metastasis.[57]2,[58]4 In the FLOT4 study, patients were randomized to receive either the FLOT regimen (5-fluorouracil, oxaliplatin, and docetaxel) or the ECF/ECX regimen (epirubicin, cisplatin, and 5-fluorouracil/capecitabine) as perioperative treatment. The results suggest that the FLOT regimen can improve the R0 resection rate (78% vs 85%) and pathological complete response rate (pCR, 6% vs 16%). In terms of long-term prognosis, the FLOT regimen led to significant improvements in overall survival (OS, 35 vs 50 months) and disease-free survival (DFS, 18 vs 30 months).[59]^2 Moreover, the RESOLVE study conducted in China showed that the SOX (S1 and oxaliplatin) regimen during the perioperative period improved 3-year DFS compared with the XELOX (capecitabine and oxaliplatin) regimen as adjuvant therapy (62.02% vs 54.78%).[60]^5 Therefore, perioperative chemotherapy for G/GEJ adenocarcinoma can increase the R0 resection rate, reduce recurrence, and improve OS. However, the prognosis of patients with locally advanced G/GEJ cancer remains unsatisfactory. In the past decade, immune checkpoint inhibitors that target programmed cell death 1 (PD-1) have been successful in the treatment of malignant tumors. Several Phase 3 clinical studies have confirmed the significant efficacy of chemotherapy combined with a PD-1 inhibitor in patients with advanced or metastatic G/GEJ cancer, and this regimen is also recommended as the standard first-line treatment.[61]6,[62]10 Phase 2 trials have also reported favorable antitumor activity with PD-1 inhibitors plus chemotherapy in patients with locally advanced G/GEJ cancer.[63]11,[64]14 The interim analysis of KEYNOTE-585, a Phase 3 trial aiming to explore the antitumor activity of neoadjuvant and adjuvant pembrolizumab (a PD-1 inhibitor) plus chemotherapy in patients with locally advanced G/GEJ adenocarcinoma, showed that perioperative pembrolizumab vs placebo improved the pCR rate (12.9% vs 2.0%, p<0.00001).[65]^15 Tumor regression after neoadjuvant therapy was significantly correlated with DFS and OS.[66]^16 The results of a study of 551 patients with G/GEJ cancer undergoing neoadjuvant treatment showed that, compared with patients with a tumor regression grade of 2 or 3, patients with a major pathological response (MPR) had a significant improvement in the 3-year OS rate (70.9% vs 48.8%).[67]^17 Overall, these studies demonstrated the efficacy of the addition of a PD-1 inhibitor for the treatment of locally advanced G/GEJ cancer, but the improvement in the pCR rate is very limited. Thus, patients who would benefit from PD-1 inhibitors combined with chemotherapy urgently need to be identified. However, biomarkers remain unclear and controversial. Moreover, the tumor immune microenvironment of locally advanced G/GEJ cancer and the effects of PD-1 inhibitors combined with chemotherapy are not fully understood. Therefore, comprehensive data analysis is needed to determine the underlying reasons and guide interventions to improve treatment efficacy. In the present study, we integrated multiomics data obtained from whole-exome sequencing (WES), whole-transcriptome sequencing (WTS), multiplex immunohistochemistry (mIHC) analysis and single-cell RNA sequencing. By comparing the genome and tumor microenvironment of MPR and non-MPR patients, we screened for effective predictive biomarkers of the response to neoadjuvant PD-1 inhibitors combined with chemotherapy. Moreover, we characterized the changes in the tumor genome and tumor microenvironment in patients before and after treatment to elucidate potential mechanisms of response and resistance. Materials and methods Data collection This study included patients with locally advanced G/GEJ cancer from a Phase 2 clinical trial ([68]NCT04908566) who had available and adequate primary tumor tissue samples for multiomics molecular analysis before and after neoadjuvant therapy. Tumor, node, metastases staging was determined by ultrasound gastroscopy and CT scan before treatment. The patients received four cycles of SOX combined with a PD-1 inhibitor, then underwent radical surgical resection and continued four cycles of SOX combined with a PD-1 inhibitor after surgery. The treatment outcome was assessed based on the Response Evaluation Criteria in Solid Tumors (RECIST) V.1.1 criteria by two independent pathologists. MPR is defined as ≤10% viable tumor cells in the resected primary tumor, excluding resected lymph nodes. We collected tumor biopsy tissues from patients before treatment for WES and WTS to profile gene expression. Furthermore, we conducted mIHC to analyze the tumor microenvironment. After radical surgical resection, surgical tissues were used to assess the MPR status, WTS was used to detect gene expression profiles, and mIHC analysis and single-cell RNA sequencing were used to analyze the tumor microenvironment and cellular heterogeneity ([69]figure 1). The method for accessing the datasets is detailed in the Data Availability Statement. Figure 1. The study design. AUC, area under curve; FOXP3, forkheadbox protein 3; MPR, major pathological response; NK, natural killer; PD-1, programmed cell death 1; Tac, activated T cells; Tcm, central memory T cells; Tem, effector memory T cells; Tex, exhausted T cells; TMB, tumor mutation burden; TPM, transcripts per million; Treg, regulatory T cell; Trm, resting memory cells; UMAP, Uniform Manifold Approximation and Projection. [70]Figure 1 [71]Open in a new tab WES and data analysis Tissue DNA was extracted from formalin-fixed paraffin-embedded (FFPE) tumor tissues using the QIAamp DNA FFPE Tissue Kit (Qiagen, Hilden, Germany) following the manufacturer’s standard protocol. Fragments between 200 and 400 bp from the sheared tissue DNA were purified (Agencourt AMPure XP Kit, Beckman Coulter, California, USA), hybridized with capture probes baits, selected with magnetic beads, and amplified. Targeted sequencing of the whole-exome region was performed using capture-based methods.[72]18,[73]20 The quality and the size of the fragments were assessed by high sensitivity DNA kit using Bioanalyzer 2100 (Agilent Technologies, California, USA). Indexed samples were sequenced on NextSeq 500 (Illumina, California, USA) with paired-end reads and an average sequencing depth of 1,000× for tissue samples. Variant calling and annotation were performed using standardized pipelines following previously established methods (Burning Rock Biotech, Guangzhou, China).[74]^18 Tumor mutation burden (TMB) per patient was computed as a ratio between the total number of non-synonymous mutations detected and the total coding region size. The mutation count included non-synonymous single nucleotide variants (SNVs) and indels detected within the coding region and ±2 bp upstream or downstream region. Copy number variations (CNVs) were analyzed based on the depth of coverage data of capture intervals. Coverage data were corrected against sequencing bias resulting from guanine cytosine (GC) content and probe design. The average coverage of all captured regions was used to normalize the coverage of different samples to comparable scales. The copy number was calculated based on the ratio between the depth of coverage in tumor samples and the average coverage of an adequate number (n>50) of samples without CNVs as references per capture